Regulators of G protein Signaling (RGS) proteins are a family of G protein-coupled receptor (GPCR) accessory proteins that are essential for the temporal and spatial control of cell signaling, including signaling downstream of the mu-opioid receptor (MOR). RGS proteins are GTPase accelerating proteins (GAPs) that accelerate the hydrolysis of Galpha bound GTP and promote the formation of inactive Galpha GDP to switch off signaling by GPCRs. We have shown that RGS proteins serve to terminate signaling of MOR to adenylate cyclase and the MAP kinase pathway. On the other hand, efficient signaling of MOR to release intracellular calcium requires RGS protein GAP activity. Thus, RGS activity controls the balance of signaling pathways within a single cell. We recently used a novel tool to explore regulation of GPCR signaling by RGS proteins: a transgenic knock-in mouse that expresses Galpha proteins that are insensitive to the GAP activity of RGS proteins. In these mice antinociception is dependent on the opioid agonist and pain assay employed. For example, in the hot-plate assay loss of RGS regulation potentiates morphine, without affecting methadone, antinociception. In contrast, in the tail-withdrawal assay removal of RGS activity decreases both morphine and methadone antinociception. This suggests the different neuronal pathways involved in these two behaviors show differential sensitivity to RGS protein action. We propose to continue our exploration of these mice to tackle a series of fundamental questions concerning MOR signaling and its relationship to antinociception. For example: Are the differences between antinociceptive tests due to site-specific variation in RGS action? What is the basis of the observed agonist differences? Can the differences be explained by effects at the level of cell signaling? What is the role of other neurotransmitter systems that also use Galpha proteins? Are more clinically-related pain models also regulated by RGS proteins? Our overall conceptual framework is as follows: 1) RGS activity controls the balance of GPCR signaling to multiple pathways and this balance may be disrupted in pain and 2) RGS-induced changes in MOR-mediated-antinociception may reflect an alteration in the balance between neurotransmitter systems, particularly the nociceptin (NOP) system. The proposed studies will advance our understanding of opioid signaling pathways and their regulation by RGS proteins and explain these actions in the context of altered behaviors. Results from this study may be exploited to develop better analgesic drugs.

Public Health Relevance

The proposed research is relevant to human health because study of the interrelationship between opioid receptors, nocicpetin receptors and G protein accessory proteins, such as RGS proteins, will increase our knowledge of the regulation of opioid signaling pathways. This in turn will provide for increased understanding of opioid-induced analgesia and other behaviors. This research is relevant to the mission of NIDA since it is designed to provide basic scientific knowledge that could lead to the better management of pain and addiction.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Project (R01)
Project #
3R01DA035316-03S1
Application #
9291217
Study Section
Program Officer
Rapaka, Rao
Project Start
2016-07-01
Project End
2019-03-31
Budget Start
2016-07-01
Budget End
2017-03-31
Support Year
3
Fiscal Year
2016
Total Cost
$55,800
Indirect Cost
$19,800
Name
University of Michigan Ann Arbor
Department
Pharmacology
Type
Schools of Medicine
DUNS #
073133571
City
Ann Arbor
State
MI
Country
United States
Zip Code
48109
McPherson, Kylie B; Leff, Emily R; Li, Ming-Hua et al. (2018) Regulators of G-Protein Signaling (RGS) Proteins Promote Receptor Coupling to G-Protein-Coupled Inwardly Rectifying Potassium (GIRK) Channels. J Neurosci 38:8737-8744
Lamberts, Jennifer T; Rosenthal, Lisa D; Jutkiewicz, Emily M et al. (2018) Role of the guanine nucleotide binding protein, G?o, in the development of morphine tolerance and dependence. Psychopharmacology (Berl) 235:71-82
Senese, Nicolas B; Oginsky, Max; Neubig, Richard R et al. (2018) Role of hippocampal 5-HT1A receptors in the antidepressant-like phenotype of mice expressing RGS-insensitive G?i2 protein. Neuropharmacology 141:296-304
Dripps, Isaac J; Boyer, Brett T; Neubig, Richard R et al. (2018) Role of signalling molecules in behaviours mediated by the ? opioid receptor agonist SNC80. Br J Pharmacol 175:891-901
Dripps, Isaac J; Wang, Qin; Neubig, Richard R et al. (2017) The role of regulator of G protein signaling 4 in delta-opioid receptor-mediated behaviors. Psychopharmacology (Berl) 234:29-39
Li, Ming-Hua; Suchland, Katherine L; Ingram, Susan L (2017) Compensatory Activation of Cannabinoid CB2 Receptor Inhibition of GABA Release in the Rostral Ventromedial Medulla in Inflammatory Pain. J Neurosci 37:626-636
Tonsfeldt, Karen J; Suchland, Katherine L; Beeson, Kathleen A et al. (2016) Sex Differences in GABAA Signaling in the Periaqueductal Gray Induced by Persistent Inflammation. J Neurosci 36:1669-81
Hillhouse, Todd M; Porter, Joseph H (2015) A brief history of the development of antidepressant drugs: from monoamines to glutamate. Exp Clin Psychopharmacol 23:1-21
Bisignano, Paola; Burford, Neil T; Shang, Yi et al. (2015) Ligand-Based Discovery of a New Scaffold for Allosteric Modulation of the ?-Opioid Receptor. J Chem Inf Model 55:1836-43
Burford, Neil T; Livingston, Kathryn E; Canals, Meritxell et al. (2015) Discovery, synthesis, and molecular pharmacology of selective positive allosteric modulators of the ?-opioid receptor. J Med Chem 58:4220-9

Showing the most recent 10 out of 11 publications